Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 499
Filtrar
1.
Circ Res ; 134(12): 1808-1823, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843289

RESUMEN

Mounting experimental and clinical evidence has revealed that adaptive immune mechanisms targeting myocardial antigens are triggered by different forms of cardiac injury and impact disease progression. B and T lymphocytes recognize specific antigens via unique adaptive immune receptors generated through a somatic rearrangement process that generates a potential repertoire of 1019 unique receptors. While the adaptive immune receptor repertoire diversity provides the basis for immunologic specificity, making sense of it can be a challenging task. In the present review, we discuss key aspects underlying the generation of TCRs (T cell receptors) and emerging tools for their study in the context of myocardial diseases. Moreover, we outline how exploring TCR repertoires could lead to a deeper understanding of myocardial pathophysiological principles and potentially serve as diagnostic tools.


Asunto(s)
Cardiomiopatías , Receptores de Antígenos de Linfocitos T , Humanos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Cardiomiopatías/inmunología , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Inmunidad Adaptativa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Miocardio/metabolismo , Miocardio/inmunología , Miocardio/patología
2.
Circ Res ; 134(12): 1824-1840, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843291

RESUMEN

Immunometabolism is an emerging field at the intersection of immunology and metabolism. Immune cell activation plays a critical role in the pathogenesis of cardiovascular diseases and is integral for regeneration during cardiac injury. We currently possess a limited understanding of the processes governing metabolic interactions between immune cells and cardiomyocytes. The impact of this intercellular crosstalk can manifest as alterations to the steady state flux of metabolites and impact cardiac contractile function. Although much of our knowledge is derived from acute inflammatory response, recent work emphasizes heterogeneity and flexibility in metabolism between cardiomyocytes and immune cells during pathological states, including ischemic, cardiometabolic, and cancer-associated disease. Metabolic adaptation is crucial because it influences immune cell activation, cytokine release, and potential therapeutic vulnerabilities. This review describes current concepts about immunometabolic regulation in the heart, focusing on intercellular crosstalk and intrinsic factors driving cellular regulation. We discuss experimental approaches to measure the cardio-immunologic crosstalk, which are necessary to uncover unknown mechanisms underlying the immune and cardiac interface. Deeper insight into these axes holds promise for therapeutic strategies that optimize cardioimmunology crosstalk for cardiac health.


Asunto(s)
Miocitos Cardíacos , Humanos , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/inmunología , Metabolismo Energético , Cardiomiopatías/metabolismo , Cardiomiopatías/inmunología , Miocardio/metabolismo , Miocardio/inmunología , Miocardio/patología
3.
J Clin Invest ; 134(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38747296

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac condition characterized by cardiac remodeling and life-threatening ventricular arrhythmias. In this issue of the JCI, Chelko, Penna, and colleagues mechanistically addressed the intricate contribution of immune-mediated injury in ACM pathogenesis. Inhibition of nuclear factor κ-B (NF-κB) and infiltration of monocyte-derived macrophages expressing C-C motif chemokine receptor-2 (CCR2) alleviated the phenotypic ACM features (i.e., fibrofatty replacement, contractile dysfunction, and ventricular arrhythmias) in desmoglein 2-mutant (Dsg2mut/mut) mice. These findings pave the way for efficacious and targetable immune therapy for patients with ACM.


Asunto(s)
Desmogleína 2 , Macrófagos , Receptores CCR2 , Animales , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Humanos , Desmogleína 2/genética , Desmogleína 2/metabolismo , Desmogleína 2/inmunología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores CCR2/antagonistas & inhibidores , FN-kappa B/metabolismo , FN-kappa B/genética , Arritmias Cardíacas/patología , Arritmias Cardíacas/inmunología , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/patología , Displasia Ventricular Derecha Arritmogénica/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/patología , Cardiomiopatías/inmunología , Cardiomiopatías/metabolismo
4.
Int Immunopharmacol ; 132: 111950, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38579564

RESUMEN

Neutrophils play a vital role in the innate immunity by perform effector functions through phagocytosis, degranulation, and forming extracellular traps. However, over-functioning of neutrophils has been associated with sterile inflammation such as Type 2 Diabetes, atherosclerosis, cancer and autoimmune disorders. Neutrophils exhibiting phenotypical and functional heterogeneity in both homeostatic and pathological conditions suggests distinct signaling pathways are activated in disease-specific stimuli and alter neutrophil functions. Hence, we examined mass spectrometry based post-translational modifications (PTM) of neutrophil proteins in response to pathologically significant stimuli, including high glucose, homocysteine and bacterial lipopolysaccharides representing diabetes-indicator, an activator of thrombosis and pathogen-associated molecule, respectively. Our data revealed that these aforesaid stimulators differentially deamidate, citrullinate, acetylate and methylate neutrophil proteins and align to distinct biological functions associated with degranulation, platelet activation, innate immune responses and metabolic alterations. The PTM patterns in response to high glucose showed an association with neutrophils extracellular traps (NETs) formation, homocysteine induced proteins PTM associated with signaling of systemic lupus erythematosus and lipopolysaccharides induced PTMs were involved in pathways related to cardiomyopathies. Our study provides novel insights into neutrophil PTM patterns and functions in response to varied pathological stimuli, which may serve as a resource to design therapeutic strategies for the management of neutrophil-centred diseases.


Asunto(s)
Trampas Extracelulares , Homocisteína , Lipopolisacáridos , Neutrófilos , Procesamiento Proteico-Postraduccional , Neutrófilos/inmunología , Neutrófilos/metabolismo , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Homocisteína/metabolismo , Glucosa/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Inmunidad Innata , Cardiomiopatías/inmunología , Cardiomiopatías/metabolismo , Transducción de Señal
5.
Circulation ; 150(1): 49-61, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38506045

RESUMEN

BACKGROUND: Viral infections can cause acute respiratory distress syndrome (ARDS), systemic inflammation, and secondary cardiovascular complications. Lung macrophage subsets change during ARDS, but the role of heart macrophages in cardiac injury during viral ARDS remains unknown. Here we investigate how immune signals typical for viral ARDS affect cardiac macrophage subsets, cardiovascular health, and systemic inflammation. METHODS: We assessed cardiac macrophage subsets using immunofluorescence histology of autopsy specimens from 21 patients with COVID-19 with SARS-CoV-2-associated ARDS and 33 patients who died from other causes. In mice, we compared cardiac immune cell dynamics after SARS-CoV-2 infection with ARDS induced by intratracheal instillation of Toll-like receptor ligands and an ACE2 (angiotensin-converting enzyme 2) inhibitor. RESULTS: In humans, SARS-CoV-2 increased total cardiac macrophage counts and led to a higher proportion of CCR2+ (C-C chemokine receptor type 2 positive) macrophages. In mice, SARS-CoV-2 and virus-free lung injury triggered profound remodeling of cardiac resident macrophages, recapitulating the clinical expansion of CCR2+ macrophages. Treating mice exposed to virus-like ARDS with a tumor necrosis factor α-neutralizing antibody reduced cardiac monocytes and inflammatory MHCIIlo CCR2+ macrophages while also preserving cardiac function. Virus-like ARDS elevated mortality in mice with pre-existing heart failure. CONCLUSIONS: Our data suggest that viral ARDS promotes cardiac inflammation by expanding the CCR2+ macrophage subset, and the associated cardiac phenotypes in mice can be elicited by activating the host immune system even without viral presence in the heart.


Asunto(s)
COVID-19 , Cardiomiopatías , Síndrome de Dificultad Respiratoria , SARS-CoV-2 , COVID-19/inmunología , COVID-19/complicaciones , COVID-19/patología , Animales , Humanos , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/virología , Ratones , Masculino , Femenino , Cardiomiopatías/inmunología , Cardiomiopatías/etiología , Cardiomiopatías/patología , Cardiomiopatías/virología , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/metabolismo , Inflamación/patología , Persona de Mediana Edad , Miocardio/patología , Miocardio/inmunología , Ratones Endogámicos C57BL , Anciano
6.
Cardiovasc Pathol ; 70: 107624, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38412903

RESUMEN

This study aimed to assess the frequency and association between transthyretin-derived (ATTR) amyloidosis and sarcoidosis in a large autopsy cohort including many cases of sudden cardiac death (SCD). We identified 73 sporadic ATTR amyloidosis cases and 11 sarcoidosis cases, among which we found two cases with concomitant ATTR amyloidosis and sarcoidosis (2.4% of all cases; 2.7% within the sporadic ATTR group). The first case involved a 92-year-old man who experienced SCD. In this patient's heart, we observed ATTR deposition and noncaseating epithelioid granulomas consistent with sarcoidosis. Focally, ATTR deposits and granulomas co-localized, with histiocyte phagocytosis of transthyretin-immunoreactive fragments. However, in most lesions, they were distributed independently. The second case was that of an 86-year-old woman who also experienced SCD. In this patient, we detected ATTR deposition in the heart and lung, while noncaseating epithelioid granulomas were only observed in the lung, liver, kidney, and thyroid. Furthermore, no co-localization of the two lesions was observed. Based on these findings, we concluded that the coexistence of ATTR amyloidosis and sarcoidosis was likely coincidental. Nevertheless, despite the rarity of the combination of these two diseases, it should be recognized as a potential cause of SCD, especially among elderly people.


Asunto(s)
Neuropatías Amiloides Familiares , Granuloma , Sarcoidosis , Humanos , Anciano de 80 o más Años , Femenino , Masculino , Granuloma/patología , Granuloma/metabolismo , Sarcoidosis/patología , Sarcoidosis/metabolismo , Sarcoidosis/complicaciones , Neuropatías Amiloides Familiares/patología , Neuropatías Amiloides Familiares/metabolismo , Neuropatías Amiloides Familiares/complicaciones , Anciano , Autopsia , Miocardio/patología , Miocardio/metabolismo , Miocardio/inmunología , Muerte Súbita Cardíaca/etiología , Muerte Súbita Cardíaca/patología , Persona de Mediana Edad , Prealbúmina/análisis , Prealbúmina/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/metabolismo , Cardiomiopatías/etiología , Cardiomiopatías/inmunología
8.
FEBS J ; 290(6): 1549-1562, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36181338

RESUMEN

Sepsis often causes cell death via pyroptosis and hence results in septic cardiomyopathy. Triggering receptors expressed in myeloid cells-1 (TREM-1) may initiate cellular cascade pathways and, in turn, induce cell death and vital organ dysfunction in sepsis, but the evidence is limited. We set to investigate the role of TREM-1 on nucleotide-binding oligomerization domain-like receptors with pyrin domain-3 (NLRP3) inflammasome activation and cardiomyocyte pyroptosis in sepsis models using cardiac cell line (HL-1) and mice. In this study, TREM-1 was found to be significantly increased in HL-1 cells challenged with lipopolysaccharide (LPS). Pyroptosis was also significantly increased in the HL-1 cells challenged with lipopolysaccharide and an NLRP3 inflammasome activator, nigericin. The close interaction between TREM-1 and structural maintenance of chromosome 4 (SMC4) was also identified. Furthermore, inhibition of TREM-1 or SMC4 prevented the upregulation of NLRP3 and decreased Gasdermin-D, IL-1ß and caspase-1 cleavage. In mice subjected to caecal ligation and puncture, the TREM-1 inhibitor LR12 decreased the expression of NLRP3 and attenuated cardiomyocyte pyroptosis, leading to improved cardiac function and prolonged survival of septic mice. Our work demonstrates that, under septic conditions, TREM-1 plays a critical role in cardiomyocyte pyroptosis. Targeting TREM-1 and its associated molecules may therefore lead to novel therapeutic treatments for septic cardiomyopathy.


Asunto(s)
Inflamasomas , Miocitos Cardíacos , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Sepsis , Receptor Activador Expresado en Células Mieloides 1 , Animales , Humanos , Ratones , Adenosina Trifosfatasas/inmunología , Cardiomiopatías/etiología , Cardiomiopatías/genética , Cardiomiopatías/inmunología , Caspasa 1/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/inmunología , Cromosomas Humanos Par 4/inmunología , Inflamasomas/agonistas , Inflamasomas/genética , Inflamasomas/inmunología , Lipopolisacáridos/efectos adversos , Lipopolisacáridos/farmacología , Células Mieloides/inmunología , Miocitos Cardíacos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/agonistas , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Piroptosis/genética , Piroptosis/inmunología , Sepsis/complicaciones , Sepsis/genética , Sepsis/inmunología , Receptor Activador Expresado en Células Mieloides 1/antagonistas & inhibidores , Receptor Activador Expresado en Células Mieloides 1/genética , Receptor Activador Expresado en Células Mieloides 1/inmunología
9.
BMC Cardiovasc Disord ; 22(1): 49, 2022 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-35152886

RESUMEN

BACKGROUND: In a Phase I study treatment with the serum amyloid P component (SAP) depleter miridesap followed by monoclonal antibody to SAP (dezamizumab) showed removal of amyloid from liver, spleen and kidney in patients with systemic amyloidosis. We report results from a Phase 2 study and concurrent immuno-positron emission tomography (PET) study assessing efficacy, pharmacodynamics, pharmacokinetics, safety and cardiac uptake (of dezamizumab) following the same intervention in patients with cardiac amyloidosis. METHODS: Both were uncontrolled open-label studies. After SAP depletion with miridesap, patients received ≤ 6 monthly doses of dezamizumab in the Phase 2 trial (n = 7), ≤ 2 doses of non-radiolabelled dezamizumab plus [89Zr]Zr-dezamizumab (total mass dose of 80 mg at session 1 and 500 mg at session 2) in the immuno-PET study (n = 2). Primary endpoints of the Phase 2 study were changed from baseline to follow-up (at 8 weeks) in left ventricular mass (LVM) by cardiac magnetic resonance imaging and safety. Primary endpoint of the immuno-PET study was [89Zr]Zr-dezamizumab cardiac uptake assessed via PET. RESULTS: Dezamizumab produced no appreciable or consistent reduction in LVM nor improvement in cardiac function in the Phase 2 study. In the immuno-PET study, measurable cardiac uptake of [89Zr]Zr-dezamizumab, although seen in both patients, was moderate to low. Uptake was notably lower in the patient with higher LVM. Treatment-associated rash with cutaneous small-vessel vasculitis was observed in both studies. Abdominal large-vessel vasculitis after initial dezamizumab dosing (300 mg) occurred in the first patient with immunoglobulin light chain amyloidosis enrolled in the Phase 2 study. Symptom resolution was nearly complete within 24 h of intravenous methylprednisolone and dezamizumab discontinuation; abdominal computed tomography imaging showed vasculitis resolution by 8 weeks. CONCLUSIONS: Unlike previous observations of visceral amyloid reduction, there was no appreciable evidence of amyloid removal in patients with cardiac amyloidosis in this Phase 2 trial, potentially related to limited cardiac uptake of dezamizumab as demonstrated in the immuno-PET study. The benefit-risk assessment for dezamizumab in cardiac amyloidosis was considered unfavourable after the incidence of large-vessel vasculitis and development for this indication was terminated. Trial registration NCT03044353 (2 February 2017) and NCT03417830 (25 January 2018).


Asunto(s)
Amiloidosis , Anticuerpos Monoclonales , Ácidos Carboxílicos , Cardiomiopatías , Tomografía de Emisión de Positrones , Pirrolidinas , Componente Amiloide P Sérico , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Amiloidosis/sangre , Amiloidosis/diagnóstico por imagen , Amiloidosis/tratamiento farmacológico , Amiloidosis/inmunología , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Ácidos Carboxílicos/efectos adversos , Ácidos Carboxílicos/uso terapéutico , Cardiomiopatías/sangre , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/inmunología , Quimioterapia Combinada , Imagen por Resonancia Magnética , Miocardio/metabolismo , Miocardio/patología , Valor Predictivo de las Pruebas , Pirrolidinas/efectos adversos , Pirrolidinas/uso terapéutico , Componente Amiloide P Sérico/antagonistas & inhibidores , Componente Amiloide P Sérico/inmunología , Factores de Tiempo , Resultado del Tratamiento , Reino Unido , Estados Unidos , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
10.
BMC Cardiovasc Disord ; 21(1): 619, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34963460

RESUMEN

OBJECTIVES: To evaluate the impact of autoantibodies against the M2-muscarinic receptor (anti-M2-R) on the clinical outcomes of patients receiving the standard treatment for peripartum cardiomyopathy (PPCM). METHODS: A total of 107 PPCM patients who received standard heart failure (HF) treatment between January 1998 and June 2020 were enrolled in this study. According to anti-M2-R reactivity, they were classified into negative (n = 59) and positive (n = 48) groups, denoted as the anti-M2-R (-) and anti-M2-R (+) groups. Echocardiography, 6-min walk distance, serum digoxin concentration (SDC), and routine laboratory tests were performed regularly for 2 years. The all-cause mortality, cardiovascular mortality, and rehospitalisation rate for HF were compared between the two groups. RESULTS: A total of 103 patients were included in the final data analysis, with 46 in the anti-M2-R (+) group and 57 in the anti-M2-R (-) group. Heart rate was lower in the anti-M2-R (+) group than in the anti-M2-R (-) group at the baseline (102.7 ± 6.1 bpm vs. 96.0 ± 6.4 bpm, p < 0.001). The initial SDC was higher in the anti-M2-R (+) group than in the anti-M2-R (-) group with the same dosage of digoxin (1.25 ± 0.45 vs. 0.78 ± 0.24 ng/mL, p < 0.001). The dosages of metoprolol and digoxin were higher in the anti-M2-R (-) patients than in the anti-M2-R (+) patients (38.8 ± 4.6 mg b.i.d. vs. 27.8 ± 5.3 mg b.i.d., p < 0.0001, respectively, for metoprolol; 0.12 ± 0.02 mg/day vs. 0.08 ± 0.04 mg/day, p < 0.0001, respectively, for digoxin). Furthermore, there was a greater improvement in cardiac function in the anti-M2-R (-) patients than in the anti-M2-R (+) patients. Multivariate analysis identified negativity for anti-M2-R as the independent predictor for the improvement of cardiac function. Rehospitalisation for HF was lower in the anti-M2-R (-) group, but all-cause mortality and cardiovascular mortality were the same. CONCLUSIONS: There were no differences in all-cause mortality or cardiovascular mortality between the two groups. Rehospitalisation rate for HF decreased in the anti-M2-R (-) group. This difference may be related to the regulation of the autonomic nervous system by anti-M2-R.


Asunto(s)
Autoanticuerpos/sangre , Sistema Nervioso Autónomo/efectos de los fármacos , Cardiomiopatías/tratamiento farmacológico , Fármacos Cardiovasculares/uso terapéutico , Corazón/inervación , Complicaciones Cardiovasculares del Embarazo/tratamiento farmacológico , Trastornos Puerperales/tratamiento farmacológico , Receptor Muscarínico M2/inmunología , Adulto , Autoinmunidad , Sistema Nervioso Autónomo/fisiopatología , Cardiomiopatías/inmunología , Cardiomiopatías/mortalidad , Cardiomiopatías/fisiopatología , Femenino , Humanos , Readmisión del Paciente , Periodo Periparto , Embarazo , Complicaciones Cardiovasculares del Embarazo/inmunología , Complicaciones Cardiovasculares del Embarazo/mortalidad , Complicaciones Cardiovasculares del Embarazo/fisiopatología , Estudios Prospectivos , Trastornos Puerperales/inmunología , Trastornos Puerperales/mortalidad , Trastornos Puerperales/fisiopatología , Recuperación de la Función , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Función Ventricular Izquierda/efectos de los fármacos
11.
Pathol Res Pract ; 227: 153635, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34653913

RESUMEN

AIM: Amyloidosis is a systemic or localized disease of protein deposition characterized by amorphous eosinophilic morphology and positivity of Congo Red staining. The typing of amyloidosis is becoming increasingly important because therapeutic agents for each amyloidosis type have been developed. Herein, the authors review the autopsy cases at an institution to reveal the putative Japanese characteristics of each amyloidosis type and evaluate the clinicopathological significance of each type. MATERIALS AND METHODS: A total of 131 autopsy cases of systemic and localized amyloidosis were retrieved for classification by immunohistochemistry. Immunohistochemistry for transthyretin, amyloid A (AA), immunoglobulin light-chain kappa and lambda, and ß2-microglobulin was performed for all cases. RESULTS: The 131 amyloidosis cases were classified as follows: 71 cases (54.2%) of transthyretin amyloidosis, 32 cases (24.4%) of AA amyloidosis, 8 cases (6.1%) of light-chain amyloidosis, and 5 cases (3.8%) of ß2-microglobulin amyloidosis, along with 15 equivocal cases (11.5%). All cases showed myocardial involvement of amyloidosis. Histopathologically, the transthyretin type was significantly associated with the interstitial and nodular patterns, and with the absence of the perivascular and endocardial patterns. The AA type was significantly associated with the perivascular and endocardial patterns, and with the absence of the nodular pattern. CONCLUSION: The authors revealed the putative characteristics of cardiac amyloidosis in Japan by using autopsy cases. About 90% of amyloidosis cases were successfully classified using only commercially available antibodies.


Asunto(s)
Amiloidosis/patología , Cardiomiopatías/patología , Inmunohistoquímica , Miocardio/patología , Adulto , Anciano , Anciano de 80 o más Años , Neuropatías Amiloides Familiares/inmunología , Neuropatías Amiloides Familiares/patología , Amiloidosis/inmunología , Autopsia , Biomarcadores/análisis , Cardiomiopatías/inmunología , Femenino , Humanos , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/inmunología , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/patología , Cadenas kappa de Inmunoglobulina/análisis , Cadenas lambda de Inmunoglobulina/análisis , Japón , Masculino , Persona de Mediana Edad , Miocardio/inmunología , Prealbúmina/análisis , Valor Predictivo de las Pruebas , Adulto Joven , Microglobulina beta-2/análisis
12.
Biomed Pharmacother ; 143: 112219, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34560540

RESUMEN

Cardiac fibrosis can lead to heart failure, arrhythmia, and sudden cardiac death, representing one of the leading causes of death due to cardiovascular diseases. Cardiac fibrosis involves several multifactorial processes that cannot be effectively controlled by the available therapies. Therefore, current research has focused on the development of novel drugs that can be used to prevent cardiac fibrosis. Recent studies on the functions of inflammasome have provided an in-depth understanding of the regulatory functions of inflammasome in cardiac fibrosis. This review summarizes the latest research on the functions of the NLRP3 inflammasome in various cardiovascular diseases. The latest findings indicate that the NLRP3 inflammasome mediates several inflammatory responses and is associated with pyroptosis, mitochondrial regulation, and myofibroblast differentiation in cardiac fibrosis. These novel findings provide insight into the vital role of the NLRP3 inflammasome in the pathogenesis of cardiac fibrosis, which can be used to identify new targets for its prevention and treatment.


Asunto(s)
Cardiomiopatías/metabolismo , Inflamasomas/metabolismo , Miocardio/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/inmunología , Cardiomiopatías/patología , Fibrosis , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/inmunología , Mediadores de Inflamación/metabolismo , Mitocondrias Cardíacas/inmunología , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Miocardio/inmunología , Miocardio/patología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Transducción de Señal
13.
Front Endocrinol (Lausanne) ; 12: 726967, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484128

RESUMEN

In March 2020, the WHO declared coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a global pandemic. Obesity was soon identified as a risk factor for poor prognosis, with an increased risk of intensive care admissions and mechanical ventilation, but also of adverse cardiovascular events. Obesity is associated with adipose tissue, chronic low-grade inflammation, and immune dysregulation with hypertrophy and hyperplasia of adipocytes and overexpression of pro-inflammatory cytokines. However, to implement appropriate therapeutic strategies, exact mechanisms must be clarified. The role of white visceral adipose tissue, increased in individuals with obesity, seems important, as a viral reservoir for SARS-CoV-2 via angiotensin-converting enzyme 2 (ACE2) receptors. After infection of host cells, the activation of pro-inflammatory cytokines creates a setting conducive to the "cytokine storm" and macrophage activation syndrome associated with progression to acute respiratory distress syndrome. In obesity, systemic viral spread, entry, and prolonged viral shedding in already inflamed adipose tissue may spur immune responses and subsequent amplification of a cytokine cascade, causing worse outcomes. More precisely, visceral adipose tissue, more than subcutaneous fat, could predict intensive care admission; and lower density of epicardial adipose tissue (EAT) could be associated with worse outcome. EAT, an ectopic adipose tissue that surrounds the myocardium, could fuel COVID-19-induced cardiac injury and myocarditis, and extensive pneumopathy, by strong expression of inflammatory mediators that could diffuse paracrinally through the vascular wall. The purpose of this review is to ascertain what mechanisms may be involved in unfavorable prognosis among COVID-19 patients with obesity, especially cardiovascular events, emphasizing the harmful role of excess ectopic adipose tissue, particularly EAT.


Asunto(s)
COVID-19/metabolismo , Cardiomiopatías/metabolismo , Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/complicaciones , COVID-19/inmunología , Cardiomiopatías/inmunología , Cardiomiopatías/patología , Cardiopatías/inmunología , Cardiopatías/metabolismo , Cardiopatías/patología , Humanos , Inflamación , Grasa Intraabdominal/patología , Obesidad/complicaciones , Obesidad/inmunología , Obesidad/patología , Pericardio , Pronóstico , SARS-CoV-2/metabolismo , Serina Endopeptidasas/metabolismo
15.
Eur J Pharmacol ; 909: 174438, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34437885

RESUMEN

Increasing evidence indicates that patients or experimental animals exposure to endotoxin (lipopolysaccharides, LPS) exert deleterious cardiac functions that greatly contribute to morbidity and mortality. The pathophysiologic processes, including NLRP3 inflammasome overactivation and cardiac inflammatory injury, are complicated. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, is a naturally occurring compound extracted from Salvia miltiorrhiza and has anti-inflammatory and cardioprotective properties. In this study we examined the effect of STS on endotoxin-induced cardiomyopathy and investigated the underlying mechanisms. An endotoxemic mouse model was established by injecting LPS (10 mg/kg). Different doses of STS were administered intraperitoneally (5, 10, or 50 mg/kg) at different time points (2/12 h, 4/12 h, and 8/12 h) after LPS challenge to assess its effect on survival of mice with endotoxemia. In parallel, cardiac function, myocardial inflammatory cytokines, cardiomyocyte pyroptosis and autophagy were evaluated to determine the extent of myocardial damage due to sepsis in the presence and absence of STS at the optimal dose (10 mg/kg) and time-point (2/12 h). The results demonstrated that STS increased the survival rates, improved the compromised cardiac function and reduced myocardial inflammatory injury associated with enhanced autophagy and mitigated NLRP3 inflammasome activation. Moreover, inhibiting of autophagy or blocking the AMPK pathway reversed STS-elicited prevention of cardiomyopathy and activated the NLRP3 inflammasome in endotoxemic mice. Collectively, our study demonstrates that STS attenuates endotoxemia-induced mortality and cardiomyopathy, which may be associated with promotion of autophagy and inhibition of NLRP3 inflammasome overactivation.


Asunto(s)
Cardiomiopatías/prevención & control , Endotoxemia/tratamiento farmacológico , Inflamasomas/antagonistas & inhibidores , Fenantrenos/farmacología , Animales , Autofagia/efectos de los fármacos , Autofagia/inmunología , Cardiomiopatías/diagnóstico , Cardiomiopatías/inmunología , Cardiomiopatías/microbiología , Modelos Animales de Enfermedad , Ecocardiografía , Endotoxemia/complicaciones , Endotoxemia/inmunología , Endotoxemia/microbiología , Endotoxinas/sangre , Endotoxinas/inmunología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/inmunología , Ventrículos Cardíacos/patología , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Masculino , Ratones , Miocitos Cardíacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fenantrenos/uso terapéutico , Piroptosis/efectos de los fármacos , Piroptosis/inmunología
16.
Cell Tissue Res ; 386(1): 79-98, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34236518

RESUMEN

The study of a desmoglein 2 murine model of arrhythmogenic cardiomyopathy revealed cardiac inflammation as a key early event leading to fibrosis. Arrhythmogenic cardiomyopathy (AC) is an inherited heart muscle disorder leading to ventricular arrhythmias and heart failure due to abnormalities in the cardiac desmosome. We examined how loss of desmoglein 2 (Dsg2) in the young murine heart leads to development of AC. Apoptosis was an early cellular phenotype, and RNA sequencing analysis revealed early activation of inflammatory-associated pathways in Dsg2-null (Dsg2-/-) hearts at postnatal day 14 (2 weeks) that were absent in the fibrotic heart of adult mice (10 weeks). This included upregulation of iRhom2/ADAM17 and its associated pro-inflammatory cytokines and receptors such as TNFα, IL6R and IL-6. Furthermore, genes linked to specific macrophage populations were also upregulated. This suggests cardiomyocyte stress triggers an early immune response to clear apoptotic cells allowing tissue remodelling later on in the fibrotic heart. Our analysis at the early disease stage suggests cardiac inflammation is an important response and may be one of the mechanisms responsible for AC disease progression.


Asunto(s)
Arritmias Cardíacas/inmunología , Cardiomiopatías/inmunología , Desmogleína 2/metabolismo , Fibrosis/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Inflamación/complicaciones , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Ratones
17.
Viruses ; 13(6)2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071557

RESUMEN

Patients with underlying cardiovascular conditions are particularly vulnerable to severe COVID-19. In this project, we aimed to characterize similarities in dysregulated immune pathways between COVID-19 patients and patients with cardiomyopathy, venous thromboembolism (VTE), or coronary artery disease (CAD). We hypothesized that these similarly dysregulated pathways may be critical to how cardiovascular diseases (CVDs) exacerbate COVID-19. To evaluate immune dysregulation in different diseases, we used four separate datasets, including RNA-sequencing data from human left ventricular cardiac muscle samples of patients with dilated or ischemic cardiomyopathy and healthy controls; RNA-sequencing data of whole blood samples from patients with single or recurrent event VTE and healthy controls; RNA-sequencing data of human peripheral blood mononuclear cells (PBMCs) from patients with and without obstructive CAD; and RNA-sequencing data of platelets from COVID-19 subjects and healthy controls. We found similar immune dysregulation profiles between patients with CVDs and COVID-19 patients. Interestingly, cardiomyopathy patients display the most similar immune landscape to COVID-19 patients. Additionally, COVID-19 patients experience greater upregulation of cytokine- and inflammasome-related genes than patients with CVDs. In all, patients with CVDs have a significant overlap of cytokine- and inflammasome-related gene expression profiles with that of COVID-19 patients, possibly explaining their greater vulnerability to severe COVID-19.


Asunto(s)
COVID-19/inmunología , COVID-19/fisiopatología , Cardiomiopatías/inmunología , Enfermedad de la Arteria Coronaria/inmunología , Tromboembolia Venosa/inmunología , COVID-19/complicaciones , COVID-19/genética , Cardiomiopatías/complicaciones , Cardiomiopatías/genética , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/genética , Citocinas/genética , Conjuntos de Datos como Asunto , Humanos , Huésped Inmunocomprometido/genética , Inflamasomas/genética , Recuento de Linfocitos , Gravedad del Paciente , RNA-Seq , Tromboembolia Venosa/complicaciones
18.
Front Immunol ; 12: 624703, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692798

RESUMEN

Accumulating evidence suggests that the breakdown of immune tolerance plays an important role in the development of myocarditis triggered by cardiotropic microbial infections. Genetic deletion of immune checkpoint molecules that are crucial for maintaining self-tolerance causes spontaneous myocarditis in mice, and cancer treatment with immune checkpoint inhibitors can induce myocarditis in humans. These results suggest that the loss of immune tolerance results in myocarditis. The tissue microenvironment influences the local immune dysregulation in autoimmunity. Recently, tenascin-C (TN-C) has been found to play a role as a local regulator of inflammation through various molecular mechanisms. TN-C is a nonstructural extracellular matrix glycoprotein expressed in the heart during early embryonic development, as well as during tissue injury or active tissue remodeling, in a spatiotemporally restricted manner. In a mouse model of autoimmune myocarditis, TN-C was detectable before inflammatory cell infiltration and myocytolysis became histologically evident; it was strongly expressed during active inflammation and disappeared with healing. TN-C activates dendritic cells to generate pathogenic autoreactive T cells and forms an important link between innate and acquired immunity.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Cardiomiopatías/metabolismo , Mediadores de Inflamación/metabolismo , Miocarditis/metabolismo , Miocardio/metabolismo , Tenascina/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Cardiomiopatías/inmunología , Cardiomiopatías/patología , Microambiente Celular , Humanos , Miocarditis/inmunología , Miocarditis/patología , Miocardio/inmunología , Miocardio/patología , Autotolerancia , Transducción de Señal
19.
Inflamm Res ; 70(4): 379-387, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33683374

RESUMEN

INTRODUCTION: Sepsis-induced myocardial dysfunction (SIMD) is a condition manifested by an intrinsic myocardial systolic and diastolic dysfunction during sepsis, which is associated with worse clinical outcomes and a higher mortality. MATERIALS AND METHODS: Several pathophysiological mechanisms including mitochondrial dysfunction, abnormal body immune reaction, metabolic reprogramming, excessive production of reactive oxygen species (ROS), and disorder of calcium regulation have been involved in SIMD. Mitophagy has potential role in protecting myocardial cells in sepsis, especially in survivors. CONCLUSION: In the current review, we focus on the role of mitochondrial dysfunction and other mitochondria-related mechanisms including immunologic imbalance, energetic reprogramming, mitophagy, and pyroptosis in the mechanisms of SIMD.


Asunto(s)
Cardiomiopatías/etiología , Mitocondrias , Sepsis/complicaciones , Animales , Cardiomiopatías/inmunología , Cardiomiopatías/metabolismo , Humanos , Tolerancia Inmunológica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Mitofagia , Miocardio , Piroptosis , Sepsis/inmunología , Sepsis/metabolismo
20.
Mol Biol Cell ; 32(7): 622-633, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33534612

RESUMEN

Dysregulation of immune responses has been linked to the generation of immunoglobulin G (IgG) autoantibodies that target human ß1ARs and contribute to deleterious cardiac outcomes. Given the benefits of ß-blockers observed in patients harboring the IgG3 subclass of autoantibodies, we investigated the role of these autoantibodies in human ß1AR function. Serum and purified IgG3(+) autoantibodies from patients with onset of cardiomyopathy were tested using human embryonic kidney (HEK) 293 cells expressing human ß1ARs. Unexpectedly, pretreatment of cells with IgG3(+) serum or purified IgG3(+) autoantibodies impaired dobutamine-mediated adenylate cyclase (AC) activity and cyclic adenosine monophosphate (cAMP) generation while enhancing biased ß-arrestin recruitment and Extracellular Regulated Kinase (ERK) activation. In contrast, the ß-blocker metoprolol increased AC activity and cAMP in the presence of IgG3(+) serum or IgG3(+) autoantibodies. Because IgG3(+) autoantibodies are specific to human ß1ARs, non-failing human hearts were used as an endogenous system to determine their ability to bias ß1AR signaling. Consistently, metoprolol increased AC activity, reflecting the ability of the IgG3(+) autoantibodies to bias ß-blocker toward G-protein coupling. Importantly, IgG3(+) autoantibodies are specific toward ß1AR as they did not alter ß2AR signaling. Thus, IgG3(+) autoantibody biases ß-blocker toward G-protein coupling while impairing agonist-mediated G-protein activation but promoting G-protein-independent ERK activation. This phenomenon may underlie the beneficial outcomes observed in patients harboring IgG3(+) ß1AR autoantibodies.


Asunto(s)
Autoanticuerpos/inmunología , Inmunoglobulina G/inmunología , Receptores Adrenérgicos beta 1/inmunología , Autoanticuerpos/sangre , Cardiomiopatías/inmunología , Cardiomiopatías/fisiopatología , AMP Cíclico , Células HEK293 , Corazón/fisiología , Humanos , Inmunoglobulina G/metabolismo , Receptores Adrenérgicos/inmunología , Receptores Adrenérgicos beta 1/metabolismo , Transducción de Señal , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...